1
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
2
|
Palomares F, Pina A, Dakhaoui H, Leiva-Castro C, Munera-Rodriguez AM, Cejudo-Guillen M, Granados B, Alba G, Santa-Maria C, Sobrino F, Lopez-Enriquez S. Dendritic Cells as a Therapeutic Strategy in Acute Myeloid Leukemia: Vaccines. Vaccines (Basel) 2024; 12:165. [PMID: 38400148 PMCID: PMC10891551 DOI: 10.3390/vaccines12020165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 01/11/2024] [Accepted: 02/04/2024] [Indexed: 02/25/2024] Open
Abstract
Dendritic cells (DCs) serve as professional antigen-presenting cells (APC) bridging innate and adaptive immunity, playing an essential role in triggering specific cellular and humoral responses against tumor and infectious antigens. Consequently, various DC-based antitumor therapeutic strategies have been developed, particularly vaccines, and have been intensively investigated specifically in the context of acute myeloid leukemia (AML). This hematological malignancy mainly affects the elderly population (those aged over 65), which usually presents a high rate of therapeutic failure and an unfavorable prognosis. In this review, we examine the current state of development and progress of vaccines in AML. The findings evidence the possible administration of DC-based vaccines as an adjuvant treatment in AML following initial therapy. Furthermore, the therapy demonstrates promising outcomes in preventing or delaying tumor relapse and exhibits synergistic effects when combined with other treatments during relapses or disease progression. On the other hand, the remarkable success observed with RNA vaccines for COVID-19, delivered in lipid nanoparticles, has revealed the efficacy and effectiveness of these types of vectors, prompting further exploration and their potential application in AML, as well as other neoplasms, loading them with tumor RNA.
Collapse
Affiliation(s)
- Francisca Palomares
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
| | - Alejandra Pina
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Hala Dakhaoui
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Camila Leiva-Castro
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Ana M. Munera-Rodriguez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Marta Cejudo-Guillen
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
- Department of Pharmacology, Pediatry, and Radiology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Beatriz Granados
- Distrito Sanitario de Atención Primaria Málaga, Sistema Sanitario Público de Andalucía, 29004 Malaga, Spain;
| | - Gonzalo Alba
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Consuelo Santa-Maria
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, 41012 Seville, Spain;
| | - Francisco Sobrino
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
| | - Soledad Lopez-Enriquez
- Department of Medical Biochemistry and Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain; (A.P.); (H.D.); (C.L.-C.); (A.M.M.-R.); (G.A.); (F.S.)
- Institute of Biomedicine of Seville (IBiS) HUVR/CSIC/University of Seville, Avda. Manuel Siurot s/n, 41013 Seville, Spain;
| |
Collapse
|
3
|
Zhao X, Zhang Z, Wen C, Huang J, Yang S, Liu J, Geng H, Peng B, Li Z, Zhang Y. The safety and anti-tumor effect of multiple peptides-pulsed dendritic cells combined with induced specific cytotoxic T lymphocytes for patients with solid tumors. Front Immunol 2023; 14:1284334. [PMID: 37942324 PMCID: PMC10628471 DOI: 10.3389/fimmu.2023.1284334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/10/2023] [Indexed: 11/10/2023] Open
Abstract
Objective The aim of this study was to explore the safety and efficacy of multiple peptide-pulsed autologous dendritic cells (DCs) combined with cytotoxic T lymphocytes (CTLs) in patients with cancer. Methods Five patients diagnosed with cancer between November 2020 and June 2021 were enrolled and received DC-CTLs therapy. Peripheral blood was collected and antigenic peptides were analyzed. The phenotype and function of DC-CTLs and the immune status of patients were detected using flow cytometry or IFN-γ ELISPOT analysis. Results DCs acquired a mature phenotype and expressed high levels of CD80, CD86, CD83, and HLA-DR after co-culture with peptides, and the DC-CTLs also exhibited high levels of IFN-γ. Peripheral blood mononuclear cells from post-treatment patients showed a stronger immune response to peptides than those prior to treatment. Importantly, four of five patients maintained a favorable immune status, of which one patient's disease-free survival lasted up to 28.2 months. No severe treatment-related adverse events were observed. Conclusion Our results show that multiple peptide-pulsed DCs combined with CTLs therapy has manageable safety and promising efficacy for cancer patients, which might provide a precise immunotherapeutic strategy for cancer.
Collapse
Affiliation(s)
- Xuan Zhao
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Zhen Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Chunli Wen
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianmin Huang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Shuangning Yang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Jinyan Liu
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
| | - Huizhen Geng
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Bing Peng
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Zibo Li
- Hebei Bio-High Technology Development Co., LTD. Shijiazhuang, Hebei, China
| | - Yi Zhang
- Biotherapy Center & Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou, Henan, China
- Henan Key Laboratory for Tumor Immunology and Biotherapy, Zhengzhou, Henan, China
- School of Life Sciences, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
4
|
Yang L, Zhuang L, Ye Z, Li L, Guan J, Gong W. Immunotherapy and biomarkers in patients with lung cancer with tuberculosis: Recent advances and future Directions. iScience 2023; 26:107881. [PMID: 37841590 PMCID: PMC10570004 DOI: 10.1016/j.isci.2023.107881] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
Lung cancer (LC) and tuberculosis (TB) are two major global public health problems, and the incidence of LC-TB is currently on the rise. Therefore effective clinical interventions are crucial for LC-TB. The aim of this review is to provide up-to-date information on the immunological profile and therapeutic biomarkers in patients with LC-TB. We discuss the immune mechanisms involved, including the immune checkpoints that play an important role in the treatment of patients with LC-TB. In addition, we explore the susceptibility of patients with LC to TB and summarise the latest research on LC-TB. Finally, we discuss future prospects in this field, including the identification of potential targets for immune intervention. In conclusion, this review provides important insights into the complex relationship between LC and TB and highlights new advances in the detection and treatment of both diseases.
Collapse
Affiliation(s)
- Ling Yang
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of PLA General Hospital, Beijing 100091, China
- Hebei North University, Zhangjiakou, Hebei 075000, China
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Li Zhuang
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Zhaoyang Ye
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Linsheng Li
- Hebei North University, Zhangjiakou, Hebei 075000, China
| | - Jingzhi Guan
- Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing 100071, China
| | - Wenping Gong
- Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, Eighth Medical Center of PLA General Hospital, Beijing 100091, China
| |
Collapse
|
5
|
Bailey H, Lee A, Eccles L, Yuan Y, Burlison H, Forshaw C, Varol N. Treatment patterns and outcomes of patients with metastatic non-small cell lung cancer in five European countries: a real-world evidence survey. BMC Cancer 2023; 23:603. [PMID: 37386452 DOI: 10.1186/s12885-023-11074-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND The landscape of non-small cell lung cancer (NSCLC) therapy is rapidly changing. This analysis aimed to understand patient characteristics, diagnosis and treatment patterns in patients with metastatic NSCLC (mNSCLC) without EGFR and ALK mutations across five European countries. METHODS Data were drawn from the Adelphi NSCLC Disease Specific Programme™, a point-in-time survey of oncologists/pulmonologists and their consulting patients in France, Germany, Italy, Spain and UK. Physicians completed record forms (RFs) for the next six consecutive consulting patients with advanced NSCLC, who then voluntarily completed questionnaires. As an oversample, physicians provided a further ten RFs specifically for patients with EGFR-wild-type mNSCLC: five patients diagnosed before March 2020 (pre-SARS-CoV-2 [COVID-19]) and five patients diagnosed from March 2020 (during COVID-19). Only EGFR-wild-type/ALK-wild-type patients were included for analysis. RESULTS Mean (standard deviation [SD]) age for 1073 patients with EGFR-wild-type/ALK-wild-type mNSCLC was 66.2 (8.9) years, 65.2% were male and 63.7% had adenocarcinoma. Level of PD-L1 expression at advanced diagnosis was < 1% for 23.1% of patients, 1-49% for 40.9% and ≥ 50% for 36.0%. Most common first-line (1L) advanced treatment was chemotherapy only (36.9%), immunotherapy monotherapy (30.5%) or immunotherapy + chemotherapy (27.6%). Of 158 patients who had progressed beyond 1L therapy, the mean (SD) time-to-treatment discontinuation was 5.1 (4.3) months; 75.9% of whom completed their 1L treatment as intended. A complete response was achieved by 6.7% and a partial response by 69.2% of patients. Of 38 patients who discontinued 1L treatment early, disease progression was reported for 73.7%. Quality of life (QoL) reported by patients was generally lower than normative reference values. Of 2373 oversample patients, physicians reported management changes for 34.7% due to COVID-19, ranging from 19.6% in Germany to 79.7% in the UK. Immunotherapy was prescribed as 1L NSCLC treatment during COVID-19 for 64.2% (n = 786) of patients and pre-COVID-19, for 47.8% (n = 549). CONCLUSIONS Real-world treatment patterns suggest that chemotherapy use remains high despite guidelines recommending immunotherapy-based 1L treatment for mNSCLC. QoL reported by patients was generally lower than population reference values. Not implying causality, 1L immunotherapy use was higher during COVID-19 than pre-COVID-19, and the UK saw the biggest impact to patient management due to COVID-19.
Collapse
Affiliation(s)
- Hollie Bailey
- Adelphi Real World, Adelphi Mill, Grimshaw Lane, Bollington, Macclesfield, Cheshire, SK10 5JB, UK
| | - Adam Lee
- Bristol Myers Squibb, Uxbridge, UK
| | | | - Yong Yuan
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Helen Burlison
- Adelphi Real World, Adelphi Mill, Grimshaw Lane, Bollington, Macclesfield, Cheshire, SK10 5JB, UK
| | - Cameron Forshaw
- Adelphi Real World, Adelphi Mill, Grimshaw Lane, Bollington, Macclesfield, Cheshire, SK10 5JB, UK.
| | | |
Collapse
|
6
|
Ye W, Li M, Luo K. Therapies Targeting Immune Cells in Tumor Microenvironment for Non-Small Cell Lung Cancer. Pharmaceutics 2023; 15:1788. [PMID: 37513975 PMCID: PMC10384189 DOI: 10.3390/pharmaceutics15071788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/02/2023] [Accepted: 06/15/2023] [Indexed: 07/30/2023] Open
Abstract
The tumor microenvironment (TME) plays critical roles in immune modulation and tumor malignancies in the process of cancer development. Immune cells constitute a significant component of the TME and influence the migration and metastasis of tumor cells. Recently, a number of therapeutic approaches targeting immune cells have proven promising and have already been used to treat different types of cancer. In particular, PD-1 and PD-L1 inhibitors have been used in the first-line setting in non-small cell lung cancer (NSCLC) with PD-L1 expression ≥1%, as approved by the FDA. In this review, we provide an introduction to the immune cells in the TME and their efficacies, and then we discuss current immunotherapies in NSCLC and scientific research progress in this field.
Collapse
Affiliation(s)
- Wei Ye
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Meiye Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
| | - Kewang Luo
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510091, China
- People's Hospital of Longhua, Affiliated Longhua People's Hospital, Southern Medical University, Shenzhen 518109, China
| |
Collapse
|
7
|
Jia G, Zhou S, Xu T, Huang Y, Li X. Conversion therapy from unresectable stage IIIC non-small-cell lung cancer to radical surgery via anti-PD-1 immunotherapy combined with chemotherapy and anti-angiogenesis: A case report and literature review. Front Oncol 2022; 12:954685. [PMID: 36185263 PMCID: PMC9515488 DOI: 10.3389/fonc.2022.954685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 08/24/2022] [Indexed: 11/25/2022] Open
Abstract
The prognosis of patients with stage IIIC non-small-cell lung cancer (NSCLC) is poor due to the loss of surgical treatment opportunities. Improving the prognosis of these patients with IIIC NSCLC urgently needs to be addressed. Here, we report a stage IIIC (T4N3M0 IIIC (AJCC 8th)) NSCLC patient treated with 2 cycles of anti-PD-1 immunotherapy combined with chemotherapy and anti-angiogenesis therapy; after two cycles of treatment, the patient achieved a partial response and obtained the opportunity for surgical treatment. After the operation, the patient achieved a pathological complete response and successfully transformed from unresectable stage IIIC lung cancer to radical surgery (ypT0N0M0). Our study is expected to provide new ideas for treating patients with unresectable stage IIIC NSCLC in the future.
Collapse
Affiliation(s)
- Guohua Jia
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shuimei Zhou
- Department of Blood Transfusion, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Tangpeng Xu
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yabing Huang
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiangpan Li
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
- *Correspondence: Xiangpan Li,
| |
Collapse
|
8
|
Nai A, Ma F, He Z, Zeng S, Bashir S, Song J, Xu M. Development and Validation of a 7-Gene Inflammatory Signature Forecasts Prognosis and Diverse Immune Landscape in Lung Adenocarcinoma. Front Mol Biosci 2022; 9:822739. [PMID: 35372503 PMCID: PMC8964604 DOI: 10.3389/fmolb.2022.822739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 02/15/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Inflammatory responses are strongly linked with tumorigenesis and cancer development. This research aimed to construct and validate a novel inflammation response–related risk predictive signature for forecasting the prognosis of patients with LUAD. Methods: Differential expression analysis, univariate Cox, LASSO, and multivariate Cox regression analyses of 200 inflammatory response–related genes (IRRG) were performed to establish a risk predictive model in the TCGA training cohort. The performance of the IRRG model was verified in eight GEO datasets. GSEA analysis, ESTIMATE algorithms, and ssGSEA analysis were applied to elucidate the possible mechanisms. Furthermore, the relationship analysis between risk score, model genes, and chemosensitivity was performed. Last, we verified the protein expression of seven model genes by immunohistochemical staining or Western blotting. Results: We constructed a novel inflammatory response–related 7-gene signature (MMP14, BTG2, LAMP3, CCL20, TLR2, IL7R, and PCDH7). Patients in the high-risk group presented markedly decreased survival time in the TCGA cohort and eight GEO cohorts than the low-risk group. Interestingly, multiple pathways related to immune response were suppressed in high-risk groups. The low infiltration levels of B cell, dendritic cell, natural killer cell, and eosinophil can significantly affect the unsatisfactory prognosis of the high-risk group in LUAD. Moreover, the tumor cells’ sensitivity to anticancer drugs was markedly related to risk scores and model genes. The protein expression of seven model genes was consistent with the mRNA expression. Conclusion: Our IRRG prognostic model can effectively forecast LUAD prognosis and is tightly related to immune infiltration.
Collapse
Affiliation(s)
- Aitao Nai
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Feng Ma
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zirui He
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuwen Zeng
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shoaib Bashir
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jian Song
- Department of Oncology, ZhongShan Torch Development Zone Hospital, Zhongshan, China
- *Correspondence: Meng Xu, ; Jian Song,
| | - Meng Xu
- Department of Oncology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Meng Xu, ; Jian Song,
| |
Collapse
|
9
|
Wieleba I, Wojas-Krawczyk K, Krawczyk P, Milanowski J. Clinical Application Perspectives of Lung Cancers 3D Tumor Microenvironment Models for In Vitro Cultures. Int J Mol Sci 2022; 23:ijms23042261. [PMID: 35216378 PMCID: PMC8876687 DOI: 10.3390/ijms23042261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the enormous progress and development of modern therapies, lung cancer remains one of the most common causes of death among men and women. The key element in the development of new anti-cancer drugs is proper planning of the preclinical research phase. The most adequate basic research exemplary for cancer study are 3D tumor microenvironment in vitro models, which allow us to avoid the use of animal models and ensure replicable culture condition. However, the question tormenting the scientist is how to choose the best tool for tumor microenvironment research, especially for extremely heterogenous lung cancer cases. In the presented review we are focused to explain the key factors of lung cancer biology, its microenvironment, and clinical gaps related to different therapies. The review summarized the most important strategies for in vitro culture models mimicking the tumor–tumor microenvironmental interaction, as well as all advantages and disadvantages were depicted. This knowledge could facilitate the right decision to designate proper pre-clinical in vitro study, based on available analytical tools and technical capabilities, to obtain more reliable and personalized results for faster introduction them into the future clinical trials.
Collapse
|
10
|
Truong CS, Yoo SY. Oncolytic Vaccinia Virus in Lung Cancer Vaccines. Vaccines (Basel) 2022; 10:240. [PMID: 35214699 PMCID: PMC8875327 DOI: 10.3390/vaccines10020240] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 11/26/2022] Open
Abstract
Therapeutic cancer vaccines represent a promising therapeutic modality via the induction of long-term immune response and reduction in adverse effects by specifically targeting tumor-associated antigens. Oncolytic virus, especially vaccinia virus (VV) is a promising cancer treatment option for effective cancer immunotherapy and thus can also be utilized in cancer vaccines. Non-small cell lung cancer (NSCLC) is likely to respond to immunotherapy, such as immune checkpoint inhibitors or cancer vaccines, since it has a high tumor mutational burden. In this review, we will summarize recent applications of VV in lung cancer treatment and discuss the potential and direction of VV-based therapeutic vaccines.
Collapse
Affiliation(s)
| | - So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Korea;
| |
Collapse
|
11
|
Fighting fire with fire: the immune system might be key in our fight against Alzheimer's disease. Drug Discov Today 2022; 27:1261-1283. [PMID: 35032668 DOI: 10.1016/j.drudis.2022.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/25/2021] [Accepted: 01/06/2022] [Indexed: 12/13/2022]
Abstract
The ultimate cause of Alzheimer's disease (AD) is still unknown and no disease-modifying treatment exists. Emerging evidence supports the concept that the immune system has a key role in AD pathogenesis. This awareness leads to the idea that specific parts of the immune system must be engaged to ward off the disease. Immunotherapy has dramatically improved the management of several previously untreatable cancers and could hold similar promise as a novel therapy for treating AD. However, before potent immunotherapies can be rationally designed as treatment against AD, we need to fully understand the dynamic interplay between AD and the different parts of our immune system. Accordingly, here we review the most important aspects of both the innate and adaptive immune system in relation to AD pathology. Teaser: Emerging results support the concept that Alzheimer's disease is affected by the inability of the immune system to contain the pathology of the brain. Here, we discuss how we can engage our immune system to fight this devastating disease.
Collapse
|